The Hpgd gene (MGI:108085) is located on mouse chromosome 8 and encodes 15-PGDH . Knockout studies reveal critical roles:
Recent studies demonstrate dual enzymatic/non-enzymatic functions in triple-negative breast cancer (TNBC):
These data suggest tumor-suppressive effects independent of prostaglandin degradation capacity .
Cancer Therapy: HPGD overexpression reduces TNBC tumor growth via KRAS/ERK pathway inhibition
Reproductive Health: Maternal HPGD deficiency causes preterm labor (gestation reduced by 12 hrs in heterozygotes)
Inflammation Control: Enzyme supplementation decreases PGE₂ levels by 80% in acute inflammation models
15-hydroxyprostaglandin dehydrogenase [NAD(+)] (EC:1.1.1.141), 15-PGDH, Hpgd, Pgdh1, Prostaglandin dehydrogenase 1.
E.coli.
MGSSHHHHHH SSGLVPRGSH MGSMHVNGKV ALVTGAAQGI GKAFAEALLL HGAKVALVDW NLEAGVKCKA ALDEQFEPQK TLFVQCDVAD QKQLRDTFRK VVDHFGRLDI LVNNAGVNNE KNWEQTLQIN LVSVISGTYL GLDYMSKQNG GEGGIIINMS SLAGLMPVAQ QPVYCASKHG IIGFTRSAAM AANLMKSGVR LNVICPGFVD TPILESIEKE ENMGQYIEYK DQIKAMMKFY GVLHPSTIAN GLINLIEDDA LNGAIMKITA SKGIHFQDYD ISPLLVKAPL TS.
HPGD encodes the NAD⁺-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH, EC 1.1.1.141), which functions as a key prostaglandin E₂ (PGE₂) catabolizing enzyme. This enzyme plays a crucial role in regulating prostaglandin levels in various tissues, affecting inflammatory responses, vascular tone, and tissue development. The significance of HPGD in research stems from its involvement in multiple physiological processes and disease conditions, including skeletal development, cardiovascular function, and neuroinflammatory disorders. Mouse models with HPGD modifications provide valuable insights into the in vivo functions of prostaglandin metabolism and its implications in disease mechanisms .
HPGD knockout mice (Hpgd⁻/⁻) exhibit several distinctive phenotypic characteristics:
Phenotype | Description | Severity |
---|---|---|
Patent ductus arteriosus | Failure of proper ductus arteriosus closure after birth | Severe |
Postnatal mortality | Early death due to high-output heart failure | High |
Prostaglandin metabolism | Elevated PGE₂ levels in tissues and circulation | Significant |
Developmental defects | Variable based on genetic background | Moderate |
Unlike human patients with HPGD mutations who develop conditions such as cranioosteoarthropathy or hypertrophic osteoarthropathy, Hpgd⁻/⁻ mice primarily manifest with patent ductus arteriosus and typically die shortly after birth due to high-output heart failure. This represents a significant species-specific difference in phenotypic expression, highlighting the importance of considering evolutionary differences when translating findings between mouse models and human disease .
Heterozygous HPGD mice (Hpgd⁺/⁻) exhibit an intermediate phenotype compared to wild-type and homozygous knockout mice. While homozygous knockouts (Hpgd⁻/⁻) typically die shortly after birth due to patent ductus arteriosus and heart failure, heterozygous mice generally survive and show more subtle alterations. These heterozygous models often display moderately elevated PGE₂ levels compared to wild-type mice but lower than homozygous knockouts. When studying HPGD-related pathways, heterozygous models offer advantages for long-term studies since they survive past the neonatal period while still exhibiting altered prostaglandin metabolism. Researchers should measure urinary and tissue PGE₂ levels to confirm the intermediate metabolic phenotype, which can serve as a biomarker for functional HPGD activity .
For reliable genotyping of HPGD mouse models, researchers should implement a comprehensive protocol that combines multiple techniques:
PCR-based genotyping: Design primers flanking the targeted region of the HPGD gene, with separate primer sets for detecting wild-type and mutant alleles.
Restriction fragment length polymorphism (RFLP) analysis: If the mutation creates or eliminates a restriction enzyme recognition site, RFLP can be used as a confirmatory method.
Quantitative PCR: For detecting gene copy number in transgenic models or confirming homozygosity/heterozygosity.
Functional validation: Measure 15-PGDH enzymatic activity in tissue samples to confirm the functional consequences of the genetic modification.
Tissue samples for genotyping can be obtained from ear punches or tail clips, with DNA extraction using standard protocols. Always include positive controls (known wild-type, heterozygous, and homozygous samples) and negative controls (no template) in genotyping reactions to ensure accuracy. For conditional knockout models, additional primers to detect the presence of Cre recombinase should be included .
When designing experiments to study neuroinflammatory responses in HPGD mouse models, researchers should implement a multi-faceted approach:
Model selection: Choose between conditional neuron-specific or microglia-specific HPGD knockout/overexpression models depending on the research question.
Experimental autoimmune encephalomyelitis (EAE) induction: For multiple sclerosis research, induce EAE using MOG₃₅₋₅₅ peptide in C57BL/6 mice and evaluate disease progression using standardized clinical scoring.
Analytical approaches:
Histopathological analysis of spinal cord and brain tissues
Flow cytometry to quantify immune cell infiltration
qRT-PCR and western blotting to assess M1/M2 microglial polarization markers
Immunohistochemistry to evaluate demyelination
In vitro validation: Complement in vivo studies with BV-2 microglial cell experiments using LPS/IL-4 treatment to induce M1/M2 polarization.
This comprehensive approach allows for detailed characterization of HPGD's role in neuroinflammation, combining behavioral, histological, and molecular analyses. When analyzing results, researchers should pay particular attention to the relationship between HPGD expression, PPARγ activation, and microglial polarization states, as these connections appear central to HPGD's role in neuroinflammatory conditions .
For accurate measurement of HPGD enzyme activity in mouse tissue samples, researchers should employ the following techniques:
Spectrophotometric NAD⁺ reduction assay: This measures the rate of NAD⁺ reduction to NADH during HPGD-catalyzed prostaglandin oxidation, detectable as an increase in absorbance at 340 nm. The assay should be performed in tissue homogenates with exogenous PGE₂ substrate and NAD⁺ cofactor.
Radiometric assay: Using ³H-labeled PGE₂ as a substrate, followed by extraction and quantification of metabolites by thin-layer chromatography or HPLC. This method offers high sensitivity but requires radioisotope handling facilities.
LC-MS/MS analysis: Measuring the conversion of PGE₂ to 15-keto-PGE₂ in tissue homogenates using liquid chromatography with tandem mass spectrometry. This offers excellent specificity and sensitivity.
For optimal results, tissue samples should be flash-frozen immediately after collection and processed on ice to preserve enzyme activity. Include appropriate controls (heat-inactivated samples, specific HPGD inhibitors) to confirm specificity. Results should be normalized to protein concentration and expressed as nmol product formed per minute per mg protein. When comparing different genetic models, consider analyzing multiple tissues as HPGD expression and activity vary significantly across organ systems .
The skeletal abnormalities observed in HPGD-deficient mice show both similarities and notable differences compared to human cranioosteoarthropathy:
Feature | Human Cranioosteoarthropathy | HPGD-deficient Mice | Significance |
---|---|---|---|
Digital clubbing | Present in all patients | Not consistently reported | Species-specific difference |
Delayed cranial suture closure | Common feature | Not typically observed | Human-specific manifestation |
Periostosis | Present in some patients | Minimal or absent | Temporal or species difference |
Joint abnormalities | Arthralgia and arthropathy | Limited data due to early mortality | Requires conditional models |
PGE₂ levels | Elevated in tissues and urine | Elevated in tissues and urine | Conserved biochemical feature |
Unlike human patients who develop pronounced skeletal abnormalities such as digital clubbing, delayed cranial suture closure, and periostosis, HPGD knockout mice primarily manifest with patent ductus arteriosus rather than overt skeletal phenotypes. This discrepancy likely reflects species-specific differences in developmental timing and tissue-specific prostaglandin metabolism. Researchers investigating cranioosteoarthropathy using mouse models should consider conditional knockout approaches or heterozygous models that survive beyond the neonatal period to better model the human skeletal phenotypes .
HPGD deficiency in mouse models results in significant cardiovascular abnormalities with potential implications for human disease:
Patent ductus arteriosus (PDA): The most striking cardiovascular phenotype in Hpgd⁻/⁻ mice is persistent patency of the ductus arteriosus, which normally closes shortly after birth. This phenotype is observed in approximately 25% of human patients with HPGD mutations, compared to 0.05% in the general population.
High-output cardiac failure: Knockout mice typically die shortly after birth due to high-output heart failure resulting from the PDA, highlighting the critical role of HPGD in postnatal cardiovascular adaptation.
Vascular tone regulation: HPGD deficiency leads to elevated PGE₂ levels, which affects vascular smooth muscle tone and can cause systemic vasodilation.
Cardiac development: While overt structural cardiac defects are uncommon, altered prostaglandin signaling may impact cardiac myocyte function and heart rate regulation.
These findings establish HPGD as a critical regulator of ductus arteriosus remodeling and postnatal cardiovascular adaptation. Mechanistically, HPGD deficiency prevents the normal postnatal decrease in prostaglandin E₂ levels that triggers ductus arteriosus closure. This connection between HPGD and ductus arteriosus patency suggests potential clinical applications in managing persistent PDA or premature ductus closure in neonates .
Differentiating between direct HPGD effects and secondary consequences of altered prostaglandin metabolism requires a strategic experimental approach:
Pharmacological interventions: Compare HPGD genetic models with wild-type mice treated with specific HPGD inhibitors versus prostaglandin receptor antagonists. Phenotypes that can be rescued by receptor antagonists but not by enzyme inhibitors likely represent secondary effects of elevated prostaglandin levels.
Prostaglandin receptor knockout crosses: Generate double knockout mice lacking both HPGD and specific prostaglandin receptors (EP1-4). Phenotypes rescued in double knockouts identify specific receptor-mediated effects.
Temporal analysis: Implement inducible knockout models to determine if acute versus chronic HPGD deficiency produces different phenotypes, helping distinguish primary from adaptive changes.
Tissue-specific conditional models: Create conditional HPGD knockouts in specific cell types to isolate direct effects in those tissues from systemic prostaglandin elevation.
Metabolomic profiling: Perform comprehensive metabolomic analysis to identify changes in multiple prostaglandin species and related metabolites, as HPGD affects several prostanoids beyond PGE₂.
This systematic approach allows researchers to deconvolute the complex consequences of HPGD deficiency, distinguishing direct enzymatic functions from secondary signaling effects mediated by elevated prostaglandin levels acting through their cognate receptors .
HPGD mouse models offer significant potential for advancing our understanding of neuroinflammatory disorders like multiple sclerosis through several key approaches:
Microglial polarization mechanisms: Recent research demonstrates that HPGD plays a critical role in regulating microglial M1/M2 polarization. HPGD overexpression inhibits M1 (pro-inflammatory) polarization while promoting M2 (anti-inflammatory) phenotypes in experimental autoimmune encephalomyelitis (EAE) models. This polarization shift represents a potential therapeutic mechanism for neuroinflammatory conditions.
Transcriptional regulation networks: HPGD expression is regulated by Nr4a1, establishing a Nr4a1-HPGD-PPARγ signaling axis that modulates neuroinflammation. Investigating this pathway in HPGD mouse models provides insights into the transcriptional control of inflammatory processes in multiple sclerosis.
Disease progression modulation: HPGD overexpression alleviates the progression of EAE, while HPGD downregulation is observed in the spinal cord of EAE mice. This suggests that targeting HPGD or its regulatory pathways could offer therapeutic approaches for multiple sclerosis.
Mechanistic studies: HPGD influences neuroinflammation by promoting PPARγ activation, a pathway with established anti-inflammatory effects. HPGD mouse models allow for detailed investigation of how prostaglandin metabolism directly impacts PPARγ signaling in neuroinflammatory conditions.
For researchers exploring these applications, combining HPGD mouse models with EAE induction provides a powerful experimental paradigm. Analysis should include clinical scoring, histopathological assessment of demyelination, and molecular characterization of microglial polarization states through M1/M2 marker expression profiles .
While the search results don't directly address HPGD's role in cancer, based on scientific knowledge about prostaglandin metabolism and inflammation in cancer development, HPGD mouse models can provide valuable insights:
Tumor suppressor function: HPGD has been identified as a potential tumor suppressor in multiple cancer types, including colorectal, breast, and lung cancers. HPGD mouse models with tissue-specific knockout or overexpression can help elucidate the mechanisms underlying this tumor suppressor function.
Inflammation-cancer connection: Given HPGD's role in regulating inflammatory prostaglandins, mouse models can help delineate how chronic inflammation driven by HPGD deficiency might contribute to cancer initiation and progression.
Therapeutic targeting approaches:
Models testing HPGD enzyme induction or stabilization
Combination approaches with COX-2 inhibitors
Investigation of downstream targets in the prostaglandin signaling pathway
Biomarker development: HPGD expression or activity levels could serve as potential biomarkers for cancer risk, progression, or treatment response, with mouse models providing validation platforms.
For cancer research applications, tissue-specific HPGD knockout models using Cre-loxP systems would be most informative, particularly when combined with established carcinogenesis protocols specific to the tissue of interest. Analysis should include tumor incidence, growth kinetics, histopathological characterization, and molecular profiling of prostaglandin metabolites and related signaling pathways.
HPGD mouse models provide valuable insights into NSAID mechanisms beyond their canonical COX inhibition:
Complementary pathway interactions: NSAIDs primarily function by inhibiting cyclooxygenase (COX) enzymes to reduce prostaglandin synthesis, while HPGD catalyzes prostaglandin degradation. HPGD mouse models allow researchers to study the interplay between these complementary regulatory mechanisms and how NSAIDs might compensate for HPGD deficiency.
Therapeutic implications in HPGD deficiency: Since HPGD mutations cause primary hypertrophic osteoarthropathy (PHO) with excessive PGE₂ levels, NSAID treatment might theoretically ameliorate symptoms by reducing prostaglandin production. Mouse models can test this therapeutic hypothesis by treating HPGD-deficient mice with various NSAIDs and monitoring clinical improvement.
Mechanistic studies on NSAID side effects: HPGD and COX pathways interact in regulating cardiovascular homeostasis. HPGD-deficient mice treated with NSAIDs can reveal how dual disruption of prostaglandin synthesis and degradation impacts cardiovascular risk, potentially explaining some NSAID-associated side effects.
Drug development applications: Understanding the relative contributions of synthesis inhibition (NSAIDs) versus enhanced degradation (HPGD activity) can guide development of novel anti-inflammatory strategies with improved safety profiles.
When designing such studies, researchers should carefully select appropriate NSAID dosing based on mouse pharmacokinetics, measure both COX activity and HPGD function, and monitor a comprehensive panel of prostaglandin metabolites to fully characterize pathway interactions .
The high neonatal mortality in HPGD knockout mice presents a significant challenge for studying adult phenotypes. Researchers can address this limitation through several strategic approaches:
Conditional knockout systems: Utilize Cre-loxP or similar systems to generate tissue-specific or inducible HPGD knockouts. This allows for normal development through the critical neonatal period before inducing HPGD deletion in adulthood.
Pharmacological intervention: Administer PGE₂ receptor antagonists during the perinatal period to temporarily block the effects of elevated PGE₂ levels, potentially increasing survival rates of HPGD-null mice.
Heterozygous models: Leverage HPGD+/- heterozygous mice, which typically survive to adulthood while still exhibiting intermediate phenotypes with moderately elevated PGE₂ levels.
Hypomorphic alleles: Generate mouse models with partially functional HPGD (hypomorphs) that retain sufficient activity to prevent neonatal lethality while still demonstrating significant reduction in enzyme function.
Ex vivo approaches: For certain studies, harvesting tissues from late-stage embryos or neonatal HPGD-null mice for ex vivo culture can provide insights into cellular phenotypes without requiring long-term survival.
When implementing these approaches, researchers should carefully document survival rates, confirm the degree of HPGD deficiency in each model, and consider potential compensatory mechanisms that might develop in conditional or hypomorphic models .
Researchers analyzing prostaglandin metabolism in HPGD mouse models frequently encounter several technical challenges:
Sample stability issues: Prostaglandins are highly labile molecules with short half-lives. Samples must be collected rapidly, processed on ice, and stabilized immediately (typically with methanol or ethanol) to prevent ex vivo degradation or production.
Baseline fluctuations: Prostaglandin levels exhibit significant biological variability based on circadian rhythms, stress, and handling. Experimental design should account for these factors through careful timing and consistent handling protocols.
Method sensitivity limitations: Endogenous prostaglandin concentrations often approach the detection limits of many assays. LC-MS/MS methods offer superior sensitivity but require specialized equipment and expertise.
Metabolite diversity: HPGD affects multiple prostaglandin species and related eicosanoids. Comprehensive metabolomic profiling rather than single-metabolite measurements provides more meaningful insights.
Tissue heterogeneity: HPGD expression varies dramatically across different cell types within a tissue. Single-cell approaches or careful microdissection may be necessary for accurate interpretation.
To address these challenges, researchers should implement standardized collection protocols, include appropriate internal standards for quantification, utilize multiple complementary analytical approaches, and always include both positive and negative controls in study designs. When interpreting results, consider the entire metabolic profile rather than isolated changes in a single prostaglandin species .
Reconciling contradictory findings between HPGD mouse models and human HPGD mutations requires careful consideration of several factors:
Species-specific developmental timing: The timing of prostaglandin-dependent developmental processes differs between mice and humans. For example, while both species require prostaglandin metabolism for ductus arteriosus closure, the relative timing and sensitivity to HPGD deficiency may differ, explaining why patent ductus arteriosus is universal in knockout mice but occurs in only 25% of human patients.
Genetic background effects: The phenotypic expression of HPGD deficiency is influenced by genetic background. Different mouse strains or human genetic backgrounds may contain modifier genes that amplify or suppress specific manifestations of HPGD deficiency.
Mutation-specific effects: Human HPGD mutations include missense, frameshift, and regulatory variants with potentially different effects on enzyme function. Replicating specific human mutations in mouse models, rather than complete knockouts, may better recapitulate human phenotypes.
Compensatory mechanisms: Mice and humans may differ in their capacity to activate compensatory pathways that partially offset HPGD deficiency. Targeted studies of related prostaglandin-metabolizing enzymes could reveal species-specific compensatory mechanisms.
Environmental factors: Environmental exposures that influence prostaglandin metabolism may differ between laboratory mice and human patients, contributing to phenotypic differences.
The future of HPGD mouse model research holds several promising directions:
Single-cell resolution studies: Implementing single-cell transcriptomics and metabolomics to understand cell-specific HPGD functions and identify previously unrecognized roles in rare cell populations.
Interaction with immune regulation: Further exploration of HPGD's role in immune cell function, particularly in microglial polarization and neuroinflammation, which could open new therapeutic avenues for neurological disorders like multiple sclerosis.
Metabolic disease connections: Investigating HPGD's potential involvement in metabolic disorders, given the emerging connections between prostaglandin signaling and metabolic regulation.
Precision disease modeling: Creating mouse models with specific human HPGD mutations to better recapitulate human disease phenotypes and enable more translational research.
Therapeutic targeting approaches: Developing methods to modulate HPGD activity or expression in a tissue-specific manner as potential therapies for conditions ranging from inflammatory disorders to cancer.
Researchers pursuing these directions should consider implementing emerging technologies such as CRISPR-Cas9 for precise genetic modifications, organoid systems for studying tissue-specific effects, and multi-omics approaches for comprehensive pathway analysis. The integration of computational modeling with experimental data will be particularly valuable for understanding the complex network of interactions between HPGD, prostaglandin signaling, and downstream effector pathways .
HPGD mouse studies have provided several valuable lessons with broad implications for prostaglandin research:
Catabolic regulation significance: While much of prostaglandin research has focused on synthesis (COX pathways), HPGD studies highlight the equal importance of catabolic regulation in determining prostaglandin bioactivity. This paradigm shift encourages balanced investigation of both synthetic and degradative pathways in eicosanoid biology.
Developmental timing precision: The lethal consequences of HPGD deficiency in mice underscore the critical timing of prostaglandin fluctuations during development. This emphasizes the need for temporal precision when studying or therapeutically targeting prostaglandin pathways.
Species-specific phenotypic expression: The differences between mouse and human HPGD-deficiency phenotypes illustrate how conserved biochemical pathways can produce divergent organismal effects. This cautions against direct extrapolation across species without validation.
Pathway interconnections: HPGD studies reveal complex interconnections between prostaglandin metabolism and other signaling networks, such as the Nr4a1-HPGD-PPARγ axis in neuroinflammation. This encourages integrated pathway analysis rather than isolated investigation of prostaglandin effects.
Therapeutic targeting potential: The distinct phenotypes of HPGD deficiency compared to COX inhibition highlight the potential for developing more selective anti-inflammatory approaches by targeting specific aspects of prostaglandin metabolism.
These insights should guide researchers to implement more nuanced, pathway-integrated approaches to prostaglandin research, with careful attention to species differences, developmental timing, and complex signaling interactions .
Emerging genetic technologies offer transformative potential for enhancing HPGD mouse model development:
CRISPR-Cas9 precision engineering: Beyond simple knockouts, CRISPR technologies enable precise introduction of specific human HPGD mutations, creation of reporter alleles, and development of conditional alleles with unprecedented efficiency. This allows direct modeling of human variants to better understand genotype-phenotype correlations.
Base and prime editing: These newer CRISPR derivatives enable precise nucleotide changes without double-strand breaks, facilitating the introduction of specific missense mutations found in human patients with minimal off-target effects.
Single-cell genomic integration: Combining single-cell RNA sequencing with HPGD mouse models allows identification of cell-specific effects and previously unrecognized HPGD functions in rare cell populations.
Spatial transcriptomics: These technologies preserve spatial information while assessing gene expression, enabling researchers to map HPGD activity and effects within complex tissue architectures.
Inducible degron systems: Rapidly inducible protein degradation systems allow temporal control of HPGD protein levels, overcoming developmental lethality while studying acute loss of function.
Tissue-specific enhancer targeting: Techniques to modify regulatory elements can produce more physiologically relevant models by altering HPGD expression patterns rather than creating complete knockouts.
15-PGDH is responsible for the oxidation of the hydroxyl group at position 15 of prostaglandins to a ketone, resulting in the loss of biological activity of these molecules . Prostaglandins are lipid compounds that have diverse hormone-like effects in animals. They are involved in various physiological processes such as inflammation, blood flow, the formation of blood clots, and the induction of labor.
The enzyme uses NAD+ (nicotinamide adenine dinucleotide) as a cofactor for its dehydrogenase activity. The reaction catalyzed by 15-PGDH is crucial for the catabolism of prostaglandins, thereby regulating their levels and activity within the body .
In mice, the HPGD gene is located on chromosome 8 . The expression of 15-PGDH is observed in various tissues, including the right lung lobe, seminal vesicle, wall of the urinary bladder, and the mucosa of the urinary bladder . This widespread expression indicates the enzyme’s significant role in multiple physiological processes.
15-PGDH has been shown to have essential roles in the prevention of early termination of pregnancy and maternal morbidity in mice . Knockout studies in mice have demonstrated that the absence of 15-PGDH leads to early pregnancy termination and maternal morbidity, highlighting its importance in reproductive health.
The recombinant form of 15-PGDH (Mouse) is produced using recombinant DNA technology, which involves inserting the HPGD gene into a suitable expression system to produce the protein in large quantities. This recombinant protein is used in various research applications to study the enzyme’s function, regulation, and potential therapeutic uses.
Mutations in the HPGD gene have been associated with several human diseases, including hypertrophic osteoarthropathy, primary autosomal recessive, and patent ductus arteriosus . The study of the mouse recombinant form of 15-PGDH provides valuable insights into the enzyme’s role in these conditions and its potential as a therapeutic target.